9
Hindawi Publishing Corporation Journal of Skin Cancer Volume 2011, Article ID 423239, 8 pages doi:10.1155/2011/423239 Review Article BRAF in Melanoma: Pathogenesis, Diagnosis, Inhibition, and Resistance Ryan J. Sullivan 1 and Keith T. Flaherty 2 1 Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114, USA 2 Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA Correspondence should be addressed to Keith T. Flaherty, kfl[email protected] Received 10 July 2011; Revised 27 September 2011; Accepted 28 September 2011 Academic Editor: S. Ugurel Copyright © 2011 R. J. Sullivan and K. T. Flaherty. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Since the initial discovery that a subset of patients with cutaneous melanoma harbor BRAF mutations, substantial research has been focused on determining the pathologic consequences of BRAF mutations, optimizing diagnostic techniques to identify these mutations, and developing therapeutic interventions to inhibit the function of this target in mutation-bearing tumors. Recently, advances have been made which are revolutionizing the standard of care for patients with BRAF mutant melanoma. This paper provides an overview on the pathogenic ramifications of mutant BRAF signaling, the latest molecular testing methods to detect BRAF mutations, and the most recent clinical data of BRAF pathway inhibitors in patients with melanoma and BRAF mutations. Finally, emerging mechanisms of resistance to BRAF inhibitors and ways of overcoming this resistance are discussed. 1. Introduction Melanoma is currently the 5th and 7th most common cancer in American men and women, respectively [1]. In addition, the incidence of melanoma has risen dramatically over the past 60 years, increasing faster than all other solid tumors [2]. Although early-stage patients can be treated successfully with surgical resection in the majority of patients, many will develop disseminated disease. The prognosis for patients with distant metastases from melanoma is dismal, and de- spite standard treatment, greater than 95% of patients with stage IV melanoma will die within five years and most pa- tients succumb within one year. More recently, preclinical discoveries have led to signif- icant advances in the understanding of the key molecular signaling events underlying the pathogenesis of melanoma. Most notably, a high percentage of tumors of melanocytic origin have been shown to harbor activating mutations of BRAF, which lead to its constitutive activity. Approximately 70–80% of acquired melanocytic nevi and 40–60% of malig- nant melanoma contain a BRAF mutation, the vast majority of which result in a single amino acid change at codon 600 (BRAF V600E )[3, 4]. The resultant unopposed, constitutive activation of extracellular signal-regulated kinase (ERK) leads to the promotion of cellular growth and opposition of apop- tosis and, ultimately, transformation into melanoma [5]. This enhanced signaling, however, also renders mutated cells susceptible to the use of small molecule inhibitors which target various BRAF pathway mediators [57]. 2. RAF Signaling and Pathogenesis of Melanoma The interaction between a growth factor receptor and its ligand typically induces a series of events, which promote cellular growth and survival. The RAS family members are GTPases which act as critical mediators in the transduction of such signals. Though RAS plays an important role in the homeostasis of normal cell turnover, death, and survival, activating mutations in RAS family members (HRAS, KRAS, and NRAS) have been identified and associated with various human malignancies [8]. In melanoma, NRAS mutations have been identified in 10–25% of tumor samples and are thought to be an important driver of oncogenesis in these patients [912]. Oncogenesis is mediated through the upregulation of several downstream signaling mechanisms,

BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

Hindawi Publishing CorporationJournal of Skin CancerVolume 2011, Article ID 423239, 8 pagesdoi:10.1155/2011/423239

Review Article

BRAF in Melanoma: Pathogenesis, Diagnosis, Inhibition,and Resistance

Ryan J. Sullivan1 and Keith T. Flaherty2

1 Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114, USA2 Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA

Correspondence should be addressed to Keith T. Flaherty, [email protected]

Received 10 July 2011; Revised 27 September 2011; Accepted 28 September 2011

Academic Editor: S. Ugurel

Copyright © 2011 R. J. Sullivan and K. T. Flaherty. This is an open access article distributed under the Creative CommonsAttribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work isproperly cited.

Since the initial discovery that a subset of patients with cutaneous melanoma harbor BRAF mutations, substantial research hasbeen focused on determining the pathologic consequences of BRAF mutations, optimizing diagnostic techniques to identify thesemutations, and developing therapeutic interventions to inhibit the function of this target in mutation-bearing tumors. Recently,advances have been made which are revolutionizing the standard of care for patients with BRAF mutant melanoma. This paperprovides an overview on the pathogenic ramifications of mutant BRAF signaling, the latest molecular testing methods to detectBRAF mutations, and the most recent clinical data of BRAF pathway inhibitors in patients with melanoma and BRAF mutations.Finally, emerging mechanisms of resistance to BRAF inhibitors and ways of overcoming this resistance are discussed.

1. Introduction

Melanoma is currently the 5th and 7th most common cancerin American men and women, respectively [1]. In addition,the incidence of melanoma has risen dramatically over thepast 60 years, increasing faster than all other solid tumors[2]. Although early-stage patients can be treated successfullywith surgical resection in the majority of patients, manywill develop disseminated disease. The prognosis for patientswith distant metastases from melanoma is dismal, and de-spite standard treatment, greater than 95% of patients withstage IV melanoma will die within five years and most pa-tients succumb within one year.

More recently, preclinical discoveries have led to signif-icant advances in the understanding of the key molecularsignaling events underlying the pathogenesis of melanoma.Most notably, a high percentage of tumors of melanocyticorigin have been shown to harbor activating mutations ofBRAF, which lead to its constitutive activity. Approximately70–80% of acquired melanocytic nevi and 40–60% of malig-nant melanoma contain a BRAF mutation, the vast majorityof which result in a single amino acid change at codon 600(BRAFV600E) [3, 4]. The resultant unopposed, constitutive

activation of extracellular signal-regulated kinase (ERK) leadsto the promotion of cellular growth and opposition of apop-tosis and, ultimately, transformation into melanoma [5].This enhanced signaling, however, also renders mutated cellssusceptible to the use of small molecule inhibitors whichtarget various BRAF pathway mediators [5–7].

2. RAF Signaling and Pathogenesis of Melanoma

The interaction between a growth factor receptor and itsligand typically induces a series of events, which promotecellular growth and survival. The RAS family members areGTPases which act as critical mediators in the transductionof such signals. Though RAS plays an important role in thehomeostasis of normal cell turnover, death, and survival,activating mutations in RAS family members (HRAS, KRAS,and NRAS) have been identified and associated with varioushuman malignancies [8]. In melanoma, NRAS mutationshave been identified in 10–25% of tumor samples andare thought to be an important driver of oncogenesis inthese patients [9–12]. Oncogenesis is mediated through theupregulation of several downstream signaling mechanisms,

Page 2: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

2 Journal of Skin Cancer

most notably the mitogen-activated protein kinase (MAPK)and the phophatidy-inositol-3-kinase (PI3K) pathways [13].

Activated RAS triggers MAPK pathway activationthrough interactions with the RAF oncoproteins (BRAF andCRAF) leading to the initiation of a progrowth signalingcascade [14]. It is unclear whether it is BRAF or CRAF thattransmits signal from mutated NRAS to MEK, but the pre-ponderance of evidence suggests that CRAF is the primarymediator [15]. RAF interacts with MAPK/ERK kinase (MEK)thereby initiating MEK phosphorylation which in turn leadsto an activating phosphorylation of ERK [14]. The activationof ERK leads to a progrowth and transforming signal, whichappears to be critical to the pathogenesis of many malig-nancies. This pathway can be initiated by either RAF iso-form, BRAF, or CRAF, though CRAF also has pro-survivaleffects, in part through the upregulation of the anti-apop-totic proteins, nuclear factor kappa B (NF-κB), and B-cellleukemia 2 (BCL-2) [14]. Interestingly, unlike CRAF, acti-vated BRAF has no other known substrates. Thus, BRAFmutant melanomas signal exclusively through MEK andsubsequently ERK leading to oncogenesis. This characteristicrenders these tumors exquisitely sensitive to potent inhibitorsof the MAPK pathway.

3. Diagnostics/Detection

Since the identification of activating mutations of BRAFin melanoma, the technology for detection has improveddramatically. Standard mutational testing for BRAF in tumortissue typically utilizes techniques such as bidirectional directfluorescent sequencing and allele-specific polymerase chainreaction which are commercially available and offer highspecificity. The sensitivity of these assays, however, is limitedin that they are only able to detect the mutation if thetumor cells constitute >5–10% of the specimen submittedfor genetic analysis [16, 17]. While this degree of sensitivityis typically sufficient to detect the presence of the BRAFV600E

mutation in a homogenous tumor nodule, this is likely notsensitive enough to detect a few tumor cells in the back-ground of a high percentage of stromal or lymphatic ele-ments, infiltrating lymphocytes, or peripheral blood cells.

One concern regarding the utilization of mutation detec-tion techniques with enhanced sensitivity is that a positivetest might actually reflect the detection of a small subsetof mutant cells. While this might have interesting scientificconsequences, the clinical relevance of a tumor containing asmall amount of mutant BRAF cells is none, as these patientswould not be expected to benefit from BRAF inhibitors.This concern is warranted, as tumor heterogeneity has beendescribed in primary melanomas [18]. In addition, whileBRAF mutations are seen in the great majority of melano-cytic nevi, vertical growth phase melanomas, and metastaticmelanoma, they are rarely detected in radial growth phasemelanomas (10%), which is thought to be the initialmalignant lesion prior to a frankly invasive lesion [19]. Thissuggests that BRAF mutation may actually be an acquiredevent in early melanoma that leads to clonal expansion andtumor progression. Such polyclonality has not been seen in

individual metastatic tumors nor when tumors across mul-tiple sites from individual patients are sampled [18, 20]. Nev-ertheless, the application of enhanced sensitivity mutationalanalysis may not be just testing tumor samples but detectingsmall numbers of representative tumor cells in a backgroundof nonmalignant cells such as in lymph nodes and peripheralblood.

More advanced techniques and assays have been devel-oped which either provide increased sensitivity or obviatethe need for increased sensitivity. These next generation testsallow for more accurate testing on samples which containonly a small amount of tumor, as well as for the detectionmutations in various peripheral blood components (i.e.,lymphocytes, mononuclear cells, plasma, serum). The utilityof many of these tests have been explored in samples frommelanoma patients with varying results.

Amplification refractory mutation systems (ARMSs) area recently described, allele-specific technique which has en-hanced sensitivity (able to detect mutation sample contain-ing 1% mutant cells) compared to standard DNA sequenc-ing of formalin fixed paraffin-embedded (FFPE) tissues[21]. Another approach which greatly enhances sensitivityfor mutation detection is the utilization of assays whichselectively amplify mutant DNA/RNA in a sample. Usinga combination of allele-specific primers and locked nucleicacid primers, the detection of 10 melanoma cells in 1 mL ofblood has been described [22]. A third approach to increasethe sensitivity of mutation detection is reported to be ableto detect one mutant cell in a thousand nonmutant cells,taking advantage of a unique restriction enzyme site inthe wild-type alleles which allows for the digestion of thewild-type alleles and thus the enrichment of the mutantalleles [23]. Finally, the incorporation of COLD-PCR leadsto a near doubling of sensitivity in the detection of BRAFmutation from FFPE tissue when using standard sequencingand pyrosequencing [24].

In addition to new technologies (ARMS) and modifica-tions to routine techniques which lead to a greater sensitivityof mutation detection, the application of standard assayson previously untested samples is also changing how weapproach BRAF testing. BRAF analysis on free DNA in theserum and plasma has been reported as has the detectionof BRAF mutations from isolated, circulating tumor cells(CTCs) [25–30]. While CTC, serum and plasma BRAFanalysis appears possible, it is yet to be determined whetherthere will be routine clinical use for one or more of theseassays or if this will remain as only an experimental approach.

While the role of standard and experimental moleculardiagnostics is being utilized to identify specific mutationsof interest (i.e., BRAFV600E), both in tissue or blood, it alsomay be worthwhile to test for other mutations and anomaliesas these may indicate sensitivity to a particular treatment.For example, Sequenom MassARRAY technology is beingused to query larger panels of oncogenic mutations, usinga primer extension reaction followed by mass spectrometryto detect the products and identify mutations with potentialclinical consequences [31, 32]. Array comparative genomehybridization (aCGH) offers the opportunity to examine theentire genome for copy number changes, including both

Page 3: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

Journal of Skin Cancer 3

amplifications and deletions that may confer sensitivity toa targeted therapy [33]. However, all these technologies areobviously limited in that they can only identify known,preselected anomalies. Whole genome analysis (WGA) hasthe potential to not only consolidate all or most of thesemodalities and tests to a single technology platform but alsoto identify additional genetic changes outside the design pa-rameters of these other assays [34]. WGA also offers the op-portunity to uncover previously unknown (perhaps patient-specific) mutations in melanoma genomes and to explorewhether particular profiles of mutations or polymorphismsmay be predictive of benefit from a particular therapy (i.e.,BRAF inhibitors, HD IL-2) [35]. Still, the clinical utility ofthese “Next Generation” tests in the care of patients withmelanoma is completely unknown.

4. Inhibitors of RAF Signaling (Less to MoreSpecific Mutant BRAF, CRAF, MEK, PerhapsMention ERK Inhibitors)

A series of small molecule inhibitors have been developedwhich target, with varying selectivity, wild-type BRAF,BRAFV600E, other mutant BRAF (at the 600 and 601 posi-tion), and CRAF. In addition, inhibitors of the downstreammediators of RAF activation, namely MEK and ERK, are alsobeing developed. In this section, only agents which have beentested clinically and reported publically are reviewed.

5. BRAF Inhibitors

5.1. Sorafenib. Sorafenib, a multitargeted tyrosine kinase in-hibitor of BRAF, CRAF, platelet-derived growth factor recep-tor (PDGFR), vascular endothelial growth factor receptor(VEGFR) 2, p38, and CKIT which was the first RAF-inhib-itor actively studied in patients with melanoma as it wasavailable for phase II testing in the same year in which BRAFmutations were first reported. Unfortunately, despite beingevaluated in numerous phase I, II, and III studies as a singleagent and in combination with chemotherapy, the clinicalutility of sorafenib has been disappointing. For example, ina single agent trial of sorafenib, the median progression-freesurvival for patients with melanoma was 11 weeks [36]. Sixpatients (16%) had stable disease at 6 months that persistedfor more than 12 months in some cases. However, only one ofthe 37 patients in the study had an actual response evaluationcriteria in solid tumor (RECIST-) defined tumor response.

This study was followed by several trials of sorafenib incombination with various cytotoxic agents, though the com-bination which was best studied was sorafenib, carboplatin,and paclitaxel [37–42]. Initial promise with this regimenwas described in a phase I trial of sorafenib in combinationwith carboplatin and paclitaxel in patients with solid tumors,where 24 patients with advanced melanoma were enrolled[39]. Ten patients with melanoma (42%) achieved anobjective response, and an additional 11 patients (46%) hadstable disease based on RECIST. The median progression-free survival was 43.7 weeks. These promising results led to aphase III trial comparing carboplatin/paclitaxel ± sorafenib

in patients with melanoma that had progressed followingtemozolomide or DTIC therapy. This trial (the PRISM study)enrolled 270 patients and showed no benefit for the additionof sorafenib to carboplatin/paclitaxel in this second-line pa-tient population [40]. The combination of carboplatin/pa-clitaxel and sorafenib was also compared to carboplatin/pa-clitaxel in a treatment naı̈ve population of patients withadvanced melanoma in a placebo-controlled randomizedphase III trial performed within the United States Intergroup(E2603). This trial enrolled 800 patients and found no benefitfor the addition of sorafenib on either median PFS or OS[41].

5.2. Higher Potency BRAF Inhibitors (PLX4032,GSK2118436).One major explanation proposed for the ineffectiveness ofsorafenib as a single agent in patients with melanoma is itsinability to completely inhibit BRAF, and in particular, BRAFcontaining the V600E mutation. Other inhibitors of BRAF,such as PLX-4032 and GSK2118436, have been developedand are more potent and selective inhibitors of mutant BRAFthan sorafenib [6, 7]. This enhanced inhibition of BRAFV600E

predictably has led to improved clinical activity of theseagents compared to sorafenib.

5.3. Vemurafenib. Vemurafenib was the first higher potencyBRAF inhibitor to complete phase I testing and show signif-icant clinical benefit [42]. In the phase I trial of PLX4032,11 of the 16 patients with tumors bearing the BRAFV600E

mutation who received a dose ≥240 mg twice daily in thedose escalation phase experienced tumor responses while noclinical responses were seen in the five patients with wild-type BRAF-containing tumor. In addition, 26 of 32 (81%)patients with the BRAFV600E mutant melanomas treated inan expansion cohort at the recommended phase II dose of960 mg twice daily had a clinical response, including twopatients achieving a complete response (CR). The estimatedmedian PFS was seven months which compares favorablyto previously available therapies for metastatic melanoma.Further, treatment with vemurafenib leads to a reduction inlevels of phosphorylated ERK (pERK) in tumors containingthe BRAFV600E mutation which is associated with clinicalresponse [43, 44]. Likely, this inhibition of pERK enhancesthe splicing of the proapoptotic BCL-2 family member BIMthereby promoting apoptosis of BRAFV600E cells [45].

These findings quickly led to the rapid accrual of botha single-agent phase II study (BRIM2) and a randomizedcontrolled phase III trial (BRIM3). The phase II trial enrolled132 patients with advanced melanoma who had received oneprior therapy. The objective response rate (ORR) was 53%with a CR rate of 5%, and the progression-free survival was6.7 months [46]. In the phase III trial, 675 patients withadvanced melanoma were randomized, to either the vemu-rafenib or dacarbazine as front-line therapy [47]. At the firstinterim analysis, treatment with vemurafenib was associatedwith a significant reduction in the risk of death and therisk of death (63% reduction) or disease progression (74%reduction), as well as a much higher ORR (48% versus 5%).These findings served as the basis for the FDA approval ofvemurafenib in August 2011.

Page 4: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

4 Journal of Skin Cancer

5.4. GSK2118436. GSK2118436 is a second higher potencyBRAF inhibitor which has shown substantial clinical activity.In a phase I/II trial, similar to PLX4032, patients with theBRAFV600E mutation treated at the two highest dose levels(150 mg twice daily and 200 mg twice daily) had a high re-sponse rate (10/16 patients, 63%) [48]. In the eight pa-tients with non-BRAFV600E mutations (V600K, V600G, andK601E) treated at a dose of ≥100 mg twice daily, three hada partial response. Both of the patients with BRAFK601E

progressed after first restaging, suggesting that only patientswith BRAF mutations at the 600 position will respond totherapy.

6. MEK Inhibitors

Inhibitors of MEK, the downstream mediator of RAF acti-vation, and the only known substrate of BRAF have shownpromise in preclinical studies in melanoma and have begunto be investigated in the clinic with some encouraging results.MEK inhibitors may also be most useful in patients withBRAFV600E mutation; as mutational status correlates stronglywith response to MEK inhibition in murine melanomaxenograft models [49].

6.1. AZD6244. Two phase I trials of AZD6244 involving pa-tients with advanced solid tumors showed this agent to bewell tolerated and to possess some antitumor activity inpatients with melanoma [50, 51]. In the first trial, threeof eight with advanced melanoma patients treated withAZD6244 achieved a partial response; BRAF and NRASmutational status was unavailable [50]. While in the secondphase I study, only one response was seen in fourteen patientswith melanoma, though this subject had a documentedBRAF mutation and a complete response ongoing for overtwo years at the time of publication [51].

In addition, AZD6244 has shown promising results inmurine models, particularly in combination with chemo-therapy, setting the stage for combination trials [52]. Build-ing upon this, a pilot study of AZD6244 in combinationdacarbazine, docetaxel, or temsirolimus in patients withadvanced melanoma was performed [53]. Eighteen patientswere treated in whom BRAF and NRAS mutational statuswas known. Clinical response was seen in five out of ninepatients (55%) with a BRAF mutation, whereas no responseswere seen in any of the nine patients without a BRAF muta-tion which included four patients with an NRAS mutation.In addition, time to progression was significantly improvedin patients with a BRAF mutation compared to those patientswithout (median 31 weeks versus 8 weeks).

6.2. GSK1120212. GSK1120212 is a reversible, selective in-hibitor of MEK1/MEK2 which has been shown, in a phase Itrial, to have single-agent efficacy in patients with advanced,BRAFV600E mutant melanoma [54]. Specifically, eight of20 patients with BRAF mutant melanoma treated withGSK2110212 had a confirmed response with two patientsachieving a CR. Interestingly, two of 22 patients with wild-type BRAF had a PR with treatment, suggesting that some

melanoma tumors are dependent on ERK/MAP kinase sig-naling despite the absence of a BRAF mutation.

6.3. PD-0325901. A phase I trial of PD-0325901 enrolled 48patients with advanced melanoma, of whom 3 (6%) had aconfirmed PR, 10 (21%) had stable disease for ≥4 months,and a total of 15 (31%) patients showed reduction in Ki-67 tumor staining [55]. Mutational analysis data of thesepatients was not provided.

6.4. AS703026. Similar results have been recently reportedwith AS703026, which is a potent MEK1/2 inhibitor. In thephase I study, three of eight patients had a partial responsewith treatment in one of two treatment schedules [56]. Muta-tional status of the melanoma patients was not reported.

While the clinical data on MEK inhibitors is encouraging,it is quite preliminary. The true value of these agentsmust await phase II and phase III trials in patients withBRAF mutant melanoma. One such trial that is currentlyongoing is a randomized, phase III trial of GSK1220212compared with chemotherapy (either dacarbazine or pacli-taxel) in patients with melanoma harboring BRAF mutations(NCIT01245062).

7. Emerging Mechanisms of Resistance toBRAF Inhibition

Importantly, it appears that the great majority of patientstreated with single agent PLX-4032 will eventually exhibitdisease progression despite successful inhibition of theBRAFV600E and a high rate of objective response early in thecourse of therapy. Preliminary studies suggest that resistanceto PLX-4032 is not related to the development of a secondmutation which impairs the binding of the treatment drug toBRAF, a resistance mechanism noted for targeted therapy inother malignancies such as nonsmall cell lung cancer, chronicmyelogenous leukemia, and gastrointestinal stromal tumor[57–59]. Instead, resistance is mediated by reactivation of theMAPK pathway in most tumors through alternative means.

It is from in vitro studies of BRAFV600E-mutated cellswhich have been generated to exhibit acquired resistance toBRAF inhibitors, that has led to the first clues as to howBRAF-mutated cells are able to survive BRAF inhibition.It appears clear that reestablishment of MAPK signaling isthe key variable in acquired resistance to BRAF inhibition[60–63]. This can be achieved through upregulation ofreceptor tyrosine kinases (i.e., PDGFRB, ERBB2) [61, 62],activation of RAS [60, 62], upregulation of CRAF [61, 63],activation of the Ser/Thr MAPK kinases (COT) [61], anddevelopment of a secondary activating mutation in MEK[64, 65]. In addition, signaling through the PI3K pathwayinitiated by insulin growth factor receptor 1 (IGF-1R) isan alternative mechanism of acquired resistance which hasalso been described [66]. Of note, each of these mechanismshave been investigated and corroborated in small numbers oftumor samples from patients who had biopsies performed atthe time of resistance, and dependence on those upregulatedor mutated signaling mediators was not demonstrated.

Page 5: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

Journal of Skin Cancer 5

Primary resistance to BRAF inhibition is seen in lessthan 10% of patients with BRAF mutant melanoma treatedwith vemurafenib [42]. While there is no data from clinicalsamples which helps to identify which patients are likelynot to benefit from BRAF inhibitors, preclinical studies sug-gest that elevated pretreatment levels of CRAF, as well as,baseline CCND1 amplification in tumors, leading to down-stream overexpression of Cyclin D1 and enhanced CDK4expression, are promising pretreatment biomarkers worthfurther investigation [63, 67].

In BRAF wild-type (BRAFWT) melanoma cells, the MAPKkinase pathway is activated by vemurafenib (and the anal-ogous PLX4720) leading to upregulation of MEK and ERKand enhanced proliferation [68, 69]. This appears to besecondary to the activation of CRAF with subsequent down-stream signaling through MEK and ERK with expected onco-genic consequences in BRAFWT cells [69]. Further, this CRAFactivation appears to be mediated through the formationof a heterodimer with the BRAFWT protein and/or CRAFhomodimer which is most apparent in RAS-mutated cells[70, 71]. In addition, PLX4720 enhances the levels of theantiapoptotic BCL-2 family member protein MCL-1 inNRAS mutant melanoma cells through enhanced signalingthrough the MAPK pathway [72]. While it is clear thatCRAF activation and enhanced MAPK pathway signalingoccur in BRAFWT melanoma cells (particularly those whichharbor an NRAS mutation) treated with BRAF inhibitorssuch as vemurafenib and PLX4720, the clinical relevance ofthis is uncertain. Specifically, it is not thought that clinicallyacquired resistance to vemurafenib, for example, occurssolely due to growth of a subset of BRAFWT melanoma cellsas persistence of the BRAFV600E mutation has been identifiedin all tumors analyzed and reported to date. In fact, it appearsthat specific changes in BRAFV600E-mutated cells allow foradaptations which lead to renewed growth despite continuedBRAF inhibition.

8. Future Directions

The establishment of single-agent efficacy of selective BRAFinhibitors, and to lesser extent MEK inhibitors, is a majorbreakthrough for the treatment of patients with BRAF muta-tion positive melanoma. Though most patients treated withthese agents are predicted to progress on treatment, theelucidation of the mechanisms of resistance described abovehelps guide future sequential and combinational therapy.Based on the finding that MAPK pathway activity is reac-tivated in melanoma following selective BRAF inhibition,the first combination trial of a selective BRAF inhibitor(GSK2118436) with a MEK inhibitor (GSK1120212) isunderway and appears tolerable with both agents being ad-ministered at their standard single-agent doses [73]. In ad-dition to this combination, trials with selective BRAF in-hibitors either in combination with or followed by IGF-R1 antagonists and other receptor tyrosine kinase inhibitorsmight be anticipated from the results of preclinical models ofresistance [61, 62, 66].

Another approach to enhancing the effectiveness of se-lective BRAF inhibitors and MEK inhibitors is the addition

of agents which might augment apoptosis. One such agentis ABT-263 which is a BH3-mimetic currently in clinicaldevelopment. In preclinical studies, the less bioavailablehomologous BH3-mimetic ABT-737 in combination witha MEK inhibitor led to enhanced lethality compared toeither agent alone [74]. Whether ABT-263 in combinationwith MEK or selective BRAF inhibitors will improve clinicaloutcomes is unknown, though it is perhaps worth exploringin an early-phase clinical trial.

In addition to combination therapy with molecularlytargeted agents which inhibit signaling induced by selectiveBRAF inhibition or promote apoptosis, another promisingapproach to maximizing the benefit of BRAF or MEK in-hibiters is to combine these agents with immunotherapy.Recently, immune checkpoint inhibitors, including the anti-CTLA-4 monoclonal antibody ipilimumab and the anti-PD monoclonal antibody MDX-1106, have shown singleagent efficacy in patients with metastatic melanoma [75, 76].Importantly, it appears that PLX4032 does not adverselyaffect human T-lymphocytes (T-cells) function while MEKinhibitors do [77]. Further, vemurafenib has been shown toimprove immune recognition by antigen-specific T cells inmelanoma [78]. These findings provide a rationale for astudy evaluating the safety and efficacy of selective BRAFinhibition in combination with immunotherapy, includingipilimumab, MDX1106, and possibly high-dose IL2.

9. Conclusions

It had been hoped for many years that the growing under-standing of the molecular pathways involved in melanomadevelopment and the increasing availability of specificinhibitors of these pathways would enable the rational de-velopment of future therapies. With the emergence of ve-murafenib and GSK2118436, the first molecularly targetedagents to lead to tumor responses in a large percentage ofpatients, a new approach to the treatment of melanoma hasbegun. As a result, all patients with advanced melanomashould have BRAF mutational analysis prior to commencingsystemic therapy. In those patients whose tumors harbor amutation in BRAF, every attempt should be made to treatthese patients with either of the two highly potent BRAFinhibitors. Additionally, as more is learned about the resis-tance mechanisms to BRAF inhibitors, the development ofcombination trials of novel molecularly targeted therapiescan be expected; further clinical improvements can only besorted out by carefully conducted preclinical and clinicalstudies that include pretreatment and on-treatment biopsies.With BRAF being established as the first point of vulnerabil-ity in melanoma, it is hoped that a molecular understandingof the limits of BRAF inhibition will lead to further clinicalbenefit.

References

[1] A. Jemal, R. Siegel, J. Xu, and E. Ward, “Cancer statistics,2010,” CA Cancer Journal for Clinicians, vol. 60, no. 5, pp. 277–300, 2010.

[2] http://seer.cancer.gov/.

Page 6: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

6 Journal of Skin Cancer

[3] H. Davies, G. R. Bignell, C. Cox et al., “Mutations of the BRAFgene in human cancer,” Nature, vol. 417, no. 6892, pp. 949–954, 2002.

[4] P. M. Pollock, U. L. Harper, K. S. Hansen et al., “High frequen-cy of BRAF mutations in nevi,” Nature Genetics, vol. 33, no. 1,pp. 19–20, 2003.

[5] A. Sharma, N. R. Trivedi, M. A. Zimmerman, D. A. Tuveson, C.D. Smith, and G. P. Robertson, “Mutant V599EB-Raf regulatesgrowth and vascular development of malignant melanomatumors,” Cancer Research, vol. 65, no. 6, pp. 2412–2421, 2005.

[6] L. A. Fecher, R. K. Amaravadi, and K. T. Flaherty, “The MAPKpathway in melanoma,” Current Opinion in Oncology, vol. 20,no. 2, pp. 183–189, 2008.

[7] J. Tsai, J. T. Lee, W. Wang et al., “Discovery of a selective in-hibitor of oncogenic B-Raf kinase with potent antimelanomaactivity,” Proceedings of the National Academy of Sciences of theUnited States of America, vol. 105, no. 8, pp. 3041–3046, 2008.

[8] J. L. Bos, “ras Oncogenes in human cancer: a review,” CancerResearch, vol. 49, no. 17, pp. 4682–4689, 1989.

[9] A. P. Albino, R. Le Strange, A. I. Oliff, M. E. Furth, and L.J. Old, “Transforming ras genes from human melanoma: amanifestation of tumour heterogeneity?” Nature, vol. 308, no.5954, pp. 69–72, 1984.

[10] N. J. Ball, J. J. Yohn, J. G. Morelli, D. A. Norris, L. E. Golitz, andJ. P. Hoeffler, “RAS mutations in human melanoma: a markerof malignant progression,” Journal of Investigative Dermatolo-gy, vol. 102, no. 3, pp. 285–290, 1994.

[11] A. Platz, U. Ringborg, E. Mansson Brahme, and B. Lagerlof,“Melanoma metastases from patients with hereditary cuta-neous malignant melanoma contain a high frequency of N-rasactivating mutations,” Melanoma Research, vol. 4, no. 3, pp.169–177, 1994.

[12] L. J. Van ’t Veer, B. M. T. Burgering, R. Versteeg et al., “N-rasmutations in human cutaneous melanoma from sun-exposedbody sites,” Molecular and Cellular Biology, vol. 9, no. 7, pp.3114–3116, 1989.

[13] A. A. Adjei, “Blocking oncogenic Ras signaling for cancer ther-apy,” Journal of the National Cancer Institute, vol. 93, no. 14,pp. 1062–1074, 2001.

[14] M. Beeram, A. Patnaik, and E. K. Rowinsky, “Raf: a strategictarget for therapeutic development against cancer,” Journal ofClinical Oncology, vol. 23, no. 27, pp. 6771–6790, 2005.

[15] N. Dumaz, R. Hayward, J. Martin et al., “In melanoma,RAS mutations are accompanied by switching signaling fromBRAF to CRAF and disrupted cyclic AMP signaling,” CancerResearch, vol. 66, no. 19, pp. 9483–9491, 2006.

[16] A. Jarry, D. Masson, E. Cassagnau, S. Parois, C. Laboisse, andM. G. Denis, “Real-time allele-specific amplification for sen-sitive detection of the BRAF mutation V600E,” Molecular andCellular Probes, vol. 18, no. 5, pp. 349–352, 2004.

[17] C. J. Miller, M. Cheung, A. Sharma et al., “Method of muta-tion analysis may contribute to discrepancies in reports ofV599EBRAF mutation frequencies in melanocytic neoplasms[1],” Journal of Investigative Dermatology, vol. 123, no. 5, pp.990–992, 2004.

[18] J. Lin, Y. Goto, H. Murata et al., “Polyclonality of BRAFmutations in primary melanoma and the selection of mutantalleles during progression,” British Journal of Cancer, vol. 104,no. 3, pp. 464–468, 2011.

[19] J. Dong, R. G. Phelps, R. Qiao et al., “BRAF oncogenic mu-tations correlate with progression rather than initiation ofhuman melanoma,” Cancer Research, vol. 63, no. 14, pp. 3883–3885, 2003.

[20] K. Omholt, A. Platz, L. Kanter, U. Ringborg, and J. Hansson,“NRAS and BRAF mutations arise early during melanomapathogenesis and are preserved throughout tumor progres-sion,” Clinical Cancer Research, vol. 9, no. 17, pp. 6483–6488,2003.

[21] G. Ellison, E. Donald, G. McWalter et al., “A comparison ofARMS and DNA sequencing for mutation analysis in clinicalbiopsy samples,” Journal of Experimental and Clinical CancerResearch, vol. 29, no. 1, article 132, 2010.

[22] R. P. Oldenburg, M. S. Liu, and M. S. Kolodney, “Selectiveamplification of rare mutations using locked nucleic acidoligonucleotides that competitively inhibit primer binding towild-type DNA,” Journal of Investigative Dermatology, vol. 128,no. 2, pp. 398–402, 2008.

[23] D. J. Panka, R. J. Sullivan, and J. W. Mier, “An inexpensive,specific and highly sensitive protocol to detect the BrafV600Emutation in melanoma tumor biopsies and blood,” MelanomaResearch, vol. 20, no. 5, pp. 401–407, 2010.

[24] P. Pinzani, C. Santucci, I. Mancini et al., “BRAFV600Edetection in melanoma is highly improved by COLD-PCR,”Clinica Chimica Acta, vol. 412, no. 11-12, pp. 901–905, 2011.

[25] M. Yancovitz, J. Yoon, M. Mikhail et al., “Detection ofmutant BRAF alleles in the plasma of patients with metastaticmelanoma,” Journal of Molecular Diagnostics, vol. 9, no. 2, pp.178–183, 2007.

[26] R. E. Board, G. Ellison, M. C. M. Orr et al., “Detection of BRAFmutations in the tumour and serum of patients enrolled inthe AZD6244 (ARRY-142886) advanced melanoma phase IIstudy,” British Journal of Cancer, vol. 101, no. 10, pp. 1724–1730, 2009.

[27] M. Shinozaki, S. J. O’Day, M. Kitago et al., “Utility of circulat-ing B-RAF DNA mutation in serum for monitoring melanomapatients receiving biochemotherapy,” Clinical Cancer Research,vol. 13, no. 7, pp. 2068–2074, 2007.

[28] M. Daniotti, V. Vallacchi, L. Rivoltini et al., “Detection ofmutated BRAFV600E variant in circulating DNA of stage III-IV melanoma patients,” International Journal of Cancer, vol.120, no. 11, pp. 2439–2444, 2007.

[29] P. Pinzani, F. Salvianti, R. Cascella et al., “Allele specificTaqman-based real-time PCR assay to quantify circulatingBRAFV600E mutated DNA in plasma of melanoma patients,”Clinica Chimica Acta, vol. 411, no. 17-18, pp. 1319–1324, 2010.

[30] M. Kitago, K. Koyanagi, T. Nakamura et al., “MRNA expres-sion and BRAF mutation in circulating melanoma cellsisolated from peripheral blood with High molecular weightmelanoma-associated antigen-specific monoclonal antibodybeads,” Clinical Chemistry, vol. 55, no. 4, pp. 757–764, 2009.

[31] S. Gabriel and L. Ziaugra, “SNP genotyping using SequenomMassARRAY 7K platform,” Current Protocols in Human Genet-ics, chapter 2, unit 2.12, 2004.

[32] S. Gabriel, L. Ziaugra, and D. Tabbaa, “SNP genotyping usingthe sequenom massARRAY iPLEX Platform,” Current Protocolsin Human Genetics, chapter 2, unit 2.12, no. 60, 2009.

[33] S. R. Moore, D. L. Persons, J. A. Sosman et al., “Detection ofcopy number alterations in metastatic melanoma by a DNAfluorescence in situ hybridization probe panel and array com-parative genomic hybridization: a southwest oncology groupstudy (S9431),” Clinical Cancer Research, vol. 14, no. 10, pp.2927–2935, 2008.

[34] M. S. Boguski, R. Arnaout, and C. Hill, “Customized care2020: how medical sequencing and network biology willenable personalized medicine,” F1000 Biology Reports, vol. 1,2009.

Page 7: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

Journal of Skin Cancer 7

[35] S. J. M. Jones, J. Laskin, Y. Y. Li et al., “Evolution of an ade-nocarcinoma in response to selection by targeted kinase in-hibitors,” Genome Biology, vol. 11, article R82, 2010.

[36] T. Eisen, T. Ahmad, K. T. Flaherty et al., “Sorafenib in ad-vanced melanoma: a Phase II randomised discontinuationtrial analysis,” British Journal of Cancer, vol. 95, no. 5, pp. 581–586, 2006.

[37] R. K. Amaravadi, L. M. Schuchter, D. F. McDermott et al.,“Phase II trial of temozolomide and sorafenib in advancedmelanoma patients with or without brain metastases,” ClinicalCancer Research, vol. 15, no. 24, pp. 7711–7718, 2009.

[38] D. F. McDermott, J. A. Sosman, R. Gonzalez et al., “Double-blind randomized phase II study of the combination ofsorafenib and dacarbazine in patients with advanced melano-ma: a report from the 11715 study group,” Journal of ClinicalOncology, vol. 26, no. 13, pp. 2178–2185, 2008.

[39] K. T. Flaherty, J. Schiller, L. M. Schuchter et al., “A Phase I trialof the oral, multikinase inhibitor sorafenib in combinationwith carboplatin and paclitaxel,” Clinical Cancer Research, vol.14, no. 15, pp. 4836–4842, 2008.

[40] A. Hauschild, S. S. Agarwala, U. Trefzer et al., “Results of aphase III, randomized, placebo-controlled study of sorafenibin combination with carboplatin and paclitaxel as second-linetreatment in patients with unresectable stage III or stage IVmelanoma,” Journal of Clinical Oncology, vol. 27, no. 17, pp.2823–2830, 2009.

[41] K. T. Flaherty, S. J. Lee, L. M. Schuchter et al., “Final results ofE2603: a double-blind, randomized phase III trial comparingcarboplatin/paclitaxel (P) with or without sorafenib (S) inmetastatic melanoma,” Journal of Clinical Oncology, vol. 28, p.8511, 2010.

[42] K. T. Flaherty, I. Puzanov, K. B. Kim et al., “Inhibition ofmutated, activated BRAF in metastatic melanoma,” New Eng-land Journal of Medicine, vol. 363, no. 9, pp. 809–819, 2010.

[43] G. Bollag, P. Hirth, J. Tsai et al., “Clinical efficacy of a RAFinhibitor needs broad target blockade in BRAF-mutant mela-noma,” Nature, vol. 467, no. 7315, pp. 596–599, 2010.

[44] E. W. Joseph, C. A. Pratilas, P. I. Poulikakos et al., “The RAFinhibitor PLX4032 inhibits ERK signaling and tumor cell pro-liferation in a V600E BRAF-selective manner,” Proceedings ofthe National Academy of Sciences of the United States of Ameri-ca, vol. 107, no. 33, pp. 14903–14908, 2010.

[45] C. C. Jiang, F. Lai, K. H. Tay et al., “Apoptosis of human mel-anoma cells induced by inhibition of B-RAF V600E involvespreferential splicing of bimS,” Cell Death and Disease, vol. 1,no. 9, article e69, 2010.

[46] A. Ribas, K. B. Kim, L. M. Schucter et al., “BRIM-2: an open-label, multicenter phase II study of vemurafenib in previ-ously treated patients with BRAF V600E mutation-positivemetastatic melanoma,” Journal of Clinical Oncology, vol. 29,2011.

[47] P. B. Chapman, A. Hauschild, C. Robert et al., “Improvedsurvival with vemurafenib in melanoma with BRAF V600Emutation,” New England Journal of Medicine, vol. 364, no. 26,pp. 2507–2516, 2011.

[48] R. Kefford, H. Arkenau, M. P. Brown et al., “Phase I/II study ofGSK2118436, a selective inhibitor of oncogenic mutant BRAFkinase, in patients with metastatic melanoma and other solidtumors,” Journal of Clinical Oncology, vol. 28, abstract 8503,2010.

[49] D. B. Solit, L. A. Garraway, C. A. Pratilas et al., “BRAFmutation predicts sensitivity to MEK inhibition,” Nature, vol.439, no. 7074, pp. 358–362, 2006.

[50] A. A. Adjei, R. B. Cohen, W. Franklin et al., “Phase I pharma-cokinetic and pharmacodynamic study of the oral, small-mol-ecule mitogen-activated protein kinase kinase 1/2 inhibitorAZD6244 (ARRY-142886) in patients with advanced cancers,”Journal of Clinical Oncology, vol. 26, no. 13, pp. 2139–2146,2008.

[51] U. Banerji, D. R. Camidge, H. M. W. Verheul et al., “The first-in-human study of the hydrogen sulfate (hyd-sulfate) capsuleof the MEK1/2 inhibitor AZD6244 (ARRY-142886): a phase Iopen-label multicenter trial in patients with advanced cancer,”Clinical Cancer Research, vol. 16, no. 5, pp. 1613–1623, 2010.

[52] N. K. Haass, K. Sproesser, T. K. Nguyen et al., “The mitogen-activated protein/extracellular signal-regulated kinase kinaseinhibitor AZD6244 (ARRY-142886) induces growth arrest inmelanoma cells and tumor regression when combined withdocetaxel,” Clinical Cancer Research, vol. 14, no. 1, pp. 230–239, 2008.

[53] S. P. Patel, A. J. Lazar, S. Mahoney et al., “Clinical responses toAZD6244 (ARRY-142886)-based combination therapy strati-fied by gene mutations in patients with metastatic melanoma,”Journal of Clinical Oncology, vol. 28, abstract 8501, 2010.

[54] J. R. Infante, L. A. Fecher, S. Nallapareddy et al., “Safety andefficacy results from the first-in-human study of the oral MEK1/2 inhibitor GSK1120212,” Journal of Clinical Oncology, vol.28, abstract 2503, 2010.

[55] P. M. LoRusso, S. S. Krishnamurthi, J. J. Rinehart et al., “PhaseI pharmacokinetic and pharmacodynamic study of the oralMAPK/ERK kinase inhibitor PD-0325901 in patients withadvanced cancers,” Clinical Cancer Research, vol. 16, no. 6, pp.1924–1937, 2010.

[56] J. Delord, N. Houede, A. Awada et al., “First-in-human phaseI safety, pharmacokinetic (PK), and pharmacodynamic (PD)analysis of the oral MEK-inhibitor AS703026 (two regimens[R]) in patients (pts) with advanced solid tumors,” Journal ofClinical Oncology, vol. 28, absrtact 2504, 2010.

[57] C. Roche-Lestienne, V. Soenen-Cornu, N. Grardel-Duflos etal., “Several types of mutations of the Abl gene can be foundin chronic myeloid leukemia patients resistant to STI571, andthey can pre-exist to the onset of treatment,” Blood, vol. 100,no. 3, pp. 1014–1018, 2002.

[58] S. Roumiantsev, N. P. Shah, M. E. Gorre et al., “Clinicalresistance to the kinase inhibitor STI-571 in chronic myeloidleukemia by mutation of Tyr-253 in the Abl kinase domain P-loop,” Proceedings of the National Academy of Sciences of theUnited States of America, vol. 99, no. 16, pp. 10700–10705,2002.

[59] E. Wardelmann, S. Merkelbach-Bruse, K. Pauls et al., “Poly-clonal evolution of multiple secondary KIT mutations in gas-trointestinal stromal tumors under treatment with imatinibmesylate,” Clinical Cancer Research, vol. 12, no. 6, pp. 1743–1749, 2006.

[60] W. D. Tap, K. W. Gong, J. Dering et al., “Pharmacodynamiccharacterization of the efficacy signals due to selective BRAFinhibition with PLX4032 in malignant melanoma,” Neoplasia,vol. 12, no. 8, pp. 637–649, 2010.

[61] C. M. Johannessen, J. S. Boehm, S. Y. Kim et al., “COT drivesresistance to RAF inhibition through MAP kinase pathway re-activation,” Nature, vol. 468, no. 7326, pp. 968–972, 2010.

[62] R. Nazarian, H. Shi, Q. Wang et al., “Melanomas acquire re-sistance to B-RAF(V600E) inhibition by RTK or N-RAS up-regulation,” Nature, vol. 468, no. 7326, pp. 973–977, 2010.

[63] C. Montagut, S. V. Sharma, T. Shioda et al., “Elevated CRAFas a potential mechanism of acquired resistance to BRAF

Page 8: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

8 Journal of Skin Cancer

inhibition in melanoma,” Cancer Research, vol. 68, no. 12, pp.4853–4861, 2008.

[64] N. Wagle, C. Emery, M. F. Berger et al., “Dissecting therapeuticresistance to RAF inhibition in melanoma by tumor genomicprofiling,” Journal of Clinical Oncology, vol. 29, pp. 3085–3096,2011.

[65] H. Wang, S. Daouti, W. -H. Li et al., “Identification of theMEK1(F129L) activating mutation as a potential mechanismof acquired resistance to MEK inhibition in human cancerscarrying the B-Raf V600E mutation,” Cancer Research, vol. 71,no. 16, pp. 5535–5545, 2011.

[66] J. Villanueva, A. Vultur, J. T. Lee et al., “Acquired resistanceto BRAF inhibitors mediated by a RAF kinase switch inmelanoma can be overcome by cotargeting MEK and IGF-1R/PI3K,” Cancer Cell, vol. 18, no. 6, pp. 683–695, 2010.

[67] K. S. M. Smalley, M. Lioni, M. D. Palma et al., “Increased cyclinD1 expression can mediate BRAF inhibitor resistance in BRAFV600E-mutated melanomas,” Molecular Cancer Therapeutics,vol. 7, no. 9, pp. 2876–2883, 2008.

[68] R. Halaban, W. Zhang, A. Bacchiocchi et al., “PLX4032,a selective BRAFV600E kinase inhibitor, activates the ERKpathway and enhances cell migration and proliferation ofBRAFWT melanoma cells,” Pigment Cell and MelanomaResearch, vol. 23, no. 2, pp. 190–200, 2010.

[69] G. Hatzivassiliou, K. Song, I. Yen et al., “RAF inhibitors primewild-type RAF to activate the MAPK pathway and enhancegrowth,” Nature, vol. 464, no. 7287, pp. 431–435, 2010.

[70] S. J. Heidorn, C. Milagre, S. Whittaker et al., “Kinase-deadBRAF and oncogenic RAS cooperate to drive tumor progres-sion through CRAF,” Cell, vol. 140, no. 2, pp. 209–221, 2010.

[71] P. I. Poulikakos, C. Zhang, G. Bollag, K. M. Shokat, and N.Rosen, “RAF inhibitors transactivate RAF dimers and ERKsignalling in cells with wild-type BRAF,” Nature, vol. 464, no.7287, pp. 427–430, 2010.

[72] F. M. Kaplan, Y. Shao, M. M. Mayberry, and A. E. Aplin, “Hy-peractivation of MEK-ERK1/2 signaling and resistanceto apoptosis induced by the oncogenic B-RAF inhibitor,PLX4720, in mutant N-RAS melanoma cells,” Oncogene, vol.30, pp. 366–371, 2011.

[73] J. R. Infante, G. S. Falchook, D. P. Lawrence et al., “PhaseI/II study to assess safety, pharmacokinetics, and efficacy ofthe oral MEK 1/2 inhibitor GSK1120212 (GSK212) dosedin combination with the oral BRAF inhibitor GSK2118436(GSK436),” Journal of Clinical Oncology, vol. 29, 2011.

[74] M. Verhaegen, J. A. Bauer, C. M. De La Vega et al., “A novelBH3 mimetic reveals a mitogen-activated protein kinase-dependent mechanism of melanoma cell death controlled byp53 and reactive oxygen species,” Cancer Research, vol. 66, no.23, pp. 11348–11359, 2006.

[75] F. S. Hodi, S. J. O’Day, D. F. McDermott et al., “Improved sur-vival with ipilimumab in patients with metastatic melanoma,”New England Journal of Medicine, vol. 363, no. 8, pp. 711–723,2010.

[76] M. Sznol, J. D. Powderly, D. C. Smith et al., “Safety andantitumor activity of biweekly MDX-1106 (Anti-PD-1, BMS-936558/ONO-4538) in patients with advanced refractorymalignancies,” Journal of Clinical Oncology, vol. 28, abstract2506, 2010.

[77] B. Comin-Anduix, T. Chodon, H. Sazegar et al., “The onco-genic BRAF kinase inhibitor PLX4032/RG7204 does not affectthe viability or function of human lymphocytes across a widerange of concentrations,” Clinical Cancer Research, vol. 16, no.24, pp. 6040–6048, 2010.

[78] A. Boni, A. P. Cogdill, P. Dang et al., “Selective BRAFV600Einhibition enhances T-cell recognition of melanoma withoutaffecting lymphocyte function,” Cancer Research, vol. 70, no.13, pp. 5213–5219, 2010.

Page 9: BRAFinMelanoma:Pathogenesis,Diagnosis,Inhibition ...downloads.hindawi.com/journals/jsc/2011/423239.pdf · Amplification refractory mutation systems (ARMSs) are a recently described,

Submit your manuscripts athttp://www.hindawi.com

Stem CellsInternational

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Disease Markers

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Immunology ResearchHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Parkinson’s Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttp://www.hindawi.com